IL1F10/IL-38 regulates immune responses by interacting with IL-36 receptor (IL-36R/IL1RL2) and modulating downstream cytokine production:
IL1F10/IL-38 acts as a "brake" on excessive inflammation by antagonizing IL-36R, which is critical in autoimmune diseases like psoriasis and inflammatory bowel disease (IBD).
IL1F10 Human His is utilized in diverse experimental settings to study immune regulation and disease mechanisms:
In murine colitis models, IL-38 knockout mice exhibit increased IL-17 expression, neutrophil infiltration, and disease activity, highlighting its protective role in gut homeostasis .
Recent studies underscore the therapeutic potential of IL1F10/IL-38 in inflammatory and autoimmune diseases:
These findings suggest IL-38 as a biomarker for IBD severity and a therapeutic candidate for inflammatory disorders.
IL1F10 (Interleukin 1 Family Member 10) is a protein-coding gene that produces a cytokine belonging to the interleukin-1 family. The protein is commonly referenced under multiple names in scientific literature, including IL-38, IL-1F10, IL-1 Theta, IL-1HY2, FIL1 Theta, and FKSG75 . When designing literature searches or comparing research findings, it's essential to incorporate all these alternative nomenclatures to ensure comprehensive coverage. The current consensus nomenclature favors IL-38 in more recent publications, though many databases and repositories still primarily list the protein under IL1F10 .
IL1F10/IL-38 functions as an immunomodulatory cytokine with primarily anti-inflammatory properties in specific contexts. Research indicates that the protein itself does not directly induce cytokine production but rather modulates existing inflammatory pathways . It has been shown to reduce IL22 and IL17A production by T-cells responding to heat-killed Candida albicans and decreases IL36G-induced production of IL8 by peripheral blood mononuclear cells . Conversely, it can increase IL6 production by dendritic cells stimulated with bacterial lipopolysaccharides (LPS) . This context-dependent immunomodulation makes IL1F10 an interesting target for studying complex immune regulation. Methodologically, researchers should consider measuring multiple cytokine outputs simultaneously when examining IL1F10 activity rather than focusing on single readouts.
Recombinant His-tagged IL1F10 protein can be produced in various expression systems, with E. coli and yeast being the most commonly documented . The choice of expression system significantly impacts protein characteristics:
E. coli expression: Provides high yield and is cost-effective but may result in proteins lacking proper folding or post-translational modifications. The recombinant proteins produced in E. coli systems typically have purity >98% as determined by reducing SDS-PAGE .
Yeast expression: Offers mammalian-like post-translational modifications while maintaining relatively high yield. Yeast-expressed IL1F10 His-tagged proteins typically achieve >90% purity as determined by SDS-PAGE .
When designing experiments, researchers should consider whether post-translational modifications are critical for their specific research questions. For structural studies, E. coli-derived protein may be sufficient, while functional studies examining receptor interactions might benefit from yeast or mammalian cell-expressed proteins.
Lyophilized His-tagged IL1F10 recombinant protein requires careful handling to maintain activity. The recommended reconstitution protocol involves dissolving the lyophilized protein in sterile PBS (pH 7.4) . Once reconstituted, the protein solution exhibits different stability profiles depending on storage temperature:
Short-term storage (2-7 days): 4-8°C is sufficient
Medium-term storage (up to 3 months): Aliquot and store at ≤-20°C
Long-term storage (up to 12 months): Store at -20°C to -80°C
To maintain protein integrity, researchers should avoid repeated freeze-thaw cycles by preparing single-use aliquots immediately after reconstitution. Additionally, it's advisable to add carrier protein (such as 0.1% BSA) when diluting to working concentrations for increased stability during experimental procedures.
Validation of His-tagged IL1F10 involves multiple complementary techniques:
Purity assessment: SDS-PAGE under reducing conditions is the standard method, with commercial preparations typically showing >90-98% purity . Western blotting using anti-His antibodies provides additional confirmation of identity.
Functional validation: Unlike many cytokines that directly induce responses, IL1F10 primarily modulates existing inflammatory pathways. Functional validation therefore requires:
Binding validation: Surface plasmon resonance or co-immunoprecipitation assays to confirm binding to IL-36R/IL1RL2
The selection of appropriate cellular and experimental models is crucial for investigating IL1F10 functions:
Cellular models:
T-cells: For studying IL1F10's effects on cytokine production (IL22, IL17A) in response to pathogens
Peripheral blood mononuclear cells (PBMCs): For examining IL1F10's modulatory effects on IL36G-induced responses
Dendritic cells: For investigating IL1F10's enhancement of LPS-stimulated IL6 production
B lymphocytes, neutrophils, monocytes: Express IL-1 family receptors and represent physiologically relevant targets
Experimental systems:
In vitro co-culture systems with mixed immune cell populations provide more physiologically relevant contexts than isolated single-cell type cultures
Ex vivo tissue explant cultures from relevant disease sites (e.g., psoriatic skin, inflammatory bowel tissues)
Mouse models with IL1F10 knockout or overexpression for in vivo studies
When designing experiments, researchers should include both positive controls (known IL-1 family cytokines) and negative controls (irrelevant recombinant proteins with similar tags) to account for potential tag-specific effects.
The presence of a histidine tag on recombinant IL1F10 introduces several methodological considerations:
Receptor binding: The position of the His-tag (N-terminal versus C-terminal) may differentially impact receptor interactions. C-terminal tags (as in many commercial preparations) generally have minimal interference with IL-36R/IL1RL2 binding, while N-terminal tags might affect the N-terminal regions potentially involved in receptor recognition .
Protein stability: His-tags may enhance protein solubility and stability in some buffer conditions but might also expose hydrophobic patches leading to aggregation over time.
Experimental artifacts: His-tags can introduce non-specific binding to metal-containing surfaces or proteins. Control experiments should include:
Comparison with tag-cleaved protein when possible
Control proteins with identical tags
Pre-blocking of potential non-specific binding sites with imidazole in some assays
To rigorously assess potential tag effects, researchers should consider comparing results from multiple versions of the protein (different tag positions or tag-free) when investigating novel IL1F10 functions.
IL1F10/IL-38 exhibits context-dependent signaling effects that require comprehensive analysis approaches:
Receptor interaction: IL1F10 acts as a ligand for IL-36R/IL1RL2, but the downstream signaling differs from other IL-1 family cytokines . Experimental approaches should examine:
Receptor binding kinetics using surface plasmon resonance
Co-immunoprecipitation of receptor complexes
FRET-based approaches to study receptor complex formation
Downstream pathway modulation:
IL1F10 reduces IL22 and IL17A production in T-cells, suggesting inhibition of pathways promoting these cytokines
IL1F10 reduces IL36G-induced IL8 production, indicating potential interference with NF-κB or MAP kinase signaling
IL1F10 increases IL6 production in dendritic cells after LPS stimulation, suggesting enhancement of specific TLR4-dependent pathways
For rigorous signaling research, phosphoproteomic approaches combined with pathway inhibitors provide the most comprehensive view of IL1F10's complex signaling effects.
The apparently contradictory effects of IL1F10 (anti-inflammatory in some contexts, pro-inflammatory in others) require sophisticated experimental approaches:
Time-course experiments: IL1F10's effects may vary temporally, with initial pro-inflammatory actions followed by resolution phases. Comprehensive time-course studies measuring multiple cytokines simultaneously are essential.
Cell-specific responses: Different cell types may respond differently to IL1F10. Single-cell analysis approaches including:
scRNA-seq of mixed populations after IL1F10 treatment
CyTOF analysis of signaling pathway activation in heterogeneous populations
Cell type-specific conditional knockout models
Dose-response considerations: IL1F10 may exhibit hormetic effects (different responses at low versus high concentrations). Full dose-response curves rather than single concentrations should be examined.
Resolution of inflammation: IL1F10 may promote resolution rather than simply inhibiting inflammation. Specialized pro-resolving mediator (SPM) analysis alongside traditional inflammatory markers provides more comprehensive understanding.
A methodological limitation in many studies is the examination of terminal readouts rather than kinetic parameters. Time-resolved approaches are crucial for understanding IL1F10's complex immunomodulatory profile.
IL1F10 has been associated with various pathological conditions that offer important research opportunities:
Disease associations:
Experimental approaches for studying pathological alterations:
Quantitative analysis of IL1F10 expression in diseased versus healthy tissues using qPCR and immunohistochemistry
Functional assessment of patient-derived IL1F10 versus recombinant protein
Genetic association studies examining IL1F10 variants and disease susceptibility
Single-cell analysis of IL1F10 expression in complex disease tissues
Methodological considerations:
Control for medication effects in patient samples
Appropriate age/sex/genetic background matching of control samples
Analysis of both protein and mRNA levels, as discrepancies may indicate post-transcriptional regulation
Single-cell approaches are particularly valuable as IL1F10 may be expressed by specific cellular subsets within complex tissues, and bulk analysis might obscure important disease-associated alterations.
Exploring IL1F10 as a therapeutic target requires specific methodological approaches:
Delivery systems for recombinant IL1F10:
PEGylation to extend half-life
Nanoparticle encapsulation for targeted delivery
Fusion proteins with tissue-targeting domains
Design of IL1F10 variants with enhanced stability or function:
Site-directed mutagenesis to identify and modify key functional residues
Development of receptor-selective mutants that retain anti-inflammatory but not pro-inflammatory properties
Creation of stabilized versions resistant to proteolytic degradation
Development of IL1F10-neutralizing approaches for conditions where inhibition is desired:
Neutralizing antibodies
Receptor antagonists
Aptamer-based inhibitors
When developing therapeutic approaches, researchers should consider implementing humanized mouse models expressing human IL1F10 and its receptors to better predict human responses, as species differences in IL-1 family members can be significant.
Distinguishing endogenous from recombinant His-tagged IL1F10 requires specific methodological considerations:
Antibody-based detection methods:
Anti-IL1F10 antibodies detect both endogenous and recombinant proteins
Anti-His tag antibodies specifically detect recombinant protein
Dual-color immunofluorescence or western blotting with both antibody types can differentiate populations
Mass spectrometry approaches:
Peptide mass fingerprinting can distinguish recombinant from endogenous proteins
Multiple reaction monitoring (MRM) assays can be developed for specific quantification of His-tagged peptides
ELISA-based detection:
A common methodological challenge is cross-reactivity with other IL-1 family members. Validation using IL1F10 knockout samples or recombinant protein standards is essential for ensuring specificity of detection methods.
Interleukin 1 Family, Member 10 (IL1F10), also known as Interleukin-38, is a protein that belongs to the interleukin 1 cytokine family. This family of cytokines plays a crucial role in the regulation of immune and inflammatory responses. IL1F10 is particularly interesting due to its immunomodulatory properties and its potential therapeutic applications.
IL1F10 is a single, non-glycosylated polypeptide chain consisting of 172 amino acids. The human recombinant version of IL1F10 is produced in Escherichia coli (E. coli) and is fused to a 20 amino acid His-tag at the N-terminus. This His-tag facilitates the purification of the protein using chromatographic techniques .
IL1F10 is expressed in various tissues, including the fetal skin, spleen, and tonsil. Within these tissues, it is generally found in the basal epithelia of the skin and in the proliferating B-cells of the tonsil . This specific expression pattern suggests that IL1F10 may have specialized roles in skin and immune system development and function.
IL1F10 is known for its immunomodulatory activity. Unlike other cytokines, IL1F10 alone does not induce cytokine production. However, it has been shown to reduce the production of IL22 and IL17A, which are cytokines involved in inflammatory responses . This suggests that IL1F10 may play a role in dampening excessive immune responses, making it a potential candidate for therapeutic interventions in inflammatory and autoimmune diseases.